Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Glia ; 71(4): 1081-1098, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36598109

RESUMEN

Astrocytes are increasingly shown to operate as an isopotential syncytium in brain function. Protoplasmic astrocytes acquire this ability to functionally go beyond the single-cell level by evolving into a spongiform morphology, cytoplasmically connecting into a syncytium, and expressing a high density of K+ conductance. However, none of these cellular/functional features exist in neonatal newborn astrocytes, which imposes a basic question of when a functional syncytium evolves in the developing brain. Our results show that the spongiform morphology of individual astrocytes and their spatial organization all reach stationary levels by postnatal day (P) 15 in the hippocampal CA1 region. Functionally, astrocytes begin to uniformly express a mature level of passive K+ conductance by P11. We next used syncytial isopotentiality measurement to monitor the maturation of the astrocyte syncytium. In uncoupled P1 astrocytes, the substitution of endogenous K+ by a Na+ -electrode solution ([Na+ ]p ) resulted in the total elimination of the physiological membrane potential (VM ), and outward K+ conductance as predicted by the Goldman-Hodgkin-Katz (GHK) equation. As more astrocytes are coupled to each other through gap junctions during development, the [Na+ ]p -induced loss of physiological VM and the outward K+ conductance is progressively compensated by the neighboring astrocytes. By P15, a stably established syncytial isopotentiality (-73 mV), and a fully compensated outward K+ conductance appeared in all [Na+ ]p -recorded astrocytes. Thus, in view of the developmental timeframe wherein a singular syncytium is anatomically and functionally established for intra-syncytium K+ equilibration, an astrocyte syncytium becomes fully operational at P15 in the mouse hippocampus.


Asunto(s)
Astrocitos , Hipocampo , Ratones , Animales , Astrocitos/fisiología , Potenciales de la Membrana/fisiología , Uniones Comunicantes/fisiología , Región CA1 Hipocampal
2.
Neurochem Res ; 48(4): 1191-1210, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35796915

RESUMEN

Now astrocytes appear to be the key contributors to the pathophysiology of major depression. Evidence in rodents shows that chronic stress is associated with a decreased expression of astrocytic GFAP-immunoreactivity within the cortex in addition to changes in the complexity and length of astrocyte processes. Furthermore, postmortem brains of individuals with depression have revealed a decrease in astrocyte density. Notably, astrocytes are extensively coupled to one another through gap junctions to form a network, or syncytium, and we have previously demonstrated that syncytial isopotentiality is a mechanism by which astrocytes function as an efficient system with respect to brain homeostasis. Interestingly, the question of how astrocyte network function changes following chronic stress is yet to be elucidated. Here, we sought to examine the effects of chronic stress on network-level astrocyte (dys)function. Using a transgenic aldh1l1-eGFP astrocyte reporter mouse, a six-week unpredictable chronic mild stress (UCMS) paradigm as a rodent model of major depression, and immunohistochemical approaches, we show that the morphology of individual astrocytes is altered by chronic stress exposure. Additionally, in astrocyte syncytial isopotentiality measurement, we found that UCMS impairs the syncytial coupling strength of astrocytes within the hippocampus and prefrontal cortex-two brain regions that have been implicated in the regulation of mood. Together, these findings reveal that chronic stress leads to astrocyte atrophy and impaired gap junction coupling, raising the prospect that both individual and network-level astrocyte functionality are important in the etiology of major depression and other neuropsychiatric disorders.


Asunto(s)
Depresión , Trastorno Depresivo Mayor , Ratones , Animales , Astrocitos/metabolismo , Encéfalo , Ratones Transgénicos , Trastorno Depresivo Mayor/metabolismo , Modelos Animales de Enfermedad , Hipocampo
3.
Glia ; 70(7): 1359-1379, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35394085

RESUMEN

Microglial control of activity-dependent plasticity and synaptic remodeling in neuronal networks has been the subject of intense research in the past several years. Although microglia-neuron interactions have been extensively studied, less is known about how microglia influence astrocyte-dependent control over neuronal structure and function. Here, we explored a role for microglia in regulating the structure and function of the astrocyte syncytium in mouse hippocampus. After depleting microglia using a CSF1R antagonist (PLX5622, Plexxikon), we observed severe disruption of astrocyte syncytial isopotentiality and dye coupling. A decrease in astrocyte-specific gap junction connexin (Cx) 30 and 43 expression, at least partially accounts for these microglia-dependent changes in astrocytes. Because neuronal function requires intact astrocyte coupling, we also evaluated the effects of microglia depletion on synaptic transmission in the hippocampus. Without microglia, the strength of synaptic transmission was reduced at baseline and after long-term potentiation (LTP). Conversely, priming microglia with systemic injections of lipopolysaccharide enhanced CA3-CA1 synaptic transmission. This microglia-induced scaling of synaptic transmission was associated with increased expression of post-synaptic scaffold proteins (Homer1) in CA1. However, astrocyte network function was not affected by microglia priming, indicating that microglia-dependent effects on astrocytes and neurons vary across functional states. Through manipulation of microglia in the brain, our results reveal the importance of microglia in homeostatic regulation of the astrocyte syncytium and scaling of synaptic transmission. These novel mechanisms uncover a new direction for future studies interrogating microglia function in various physiological and pathological contexts.


Asunto(s)
Astrocitos , Microglía , Animales , Astrocitos/metabolismo , Conexina 30/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Ratones , Microglía/metabolismo , Plasticidad Neuronal/fisiología , Transmisión Sináptica/fisiología
4.
Prog Neurobiol ; 213: 102264, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35283239

RESUMEN

The complexity of astrocyte morphology and syncytial coupling through gap junctions are crucial for astrocyte function in the brain. However, the ultrastructural details of astrocyte arborization and interactions between neighboring astrocytes remain unknown. While a prevailing view is that synapses selectively contact peripheral astrocyte processes, the precise spatial-location selectivity of synapses abutting astrocytes is unresolved. Additionally, knowing the location and quantity of vesicles and mitochondria are prerequisites to answer two emerging questions - whether astrocytes have a signaling role within the brain and whether astrocytes are highly metabolically active. Here, we provided structural context for these questions by tracing and 3D reconstructing three neighboring astrocytes using serial block-face scanning electron microscopy. Our reconstructions reveal a spongiform astrocytic morphology resulting from the abundance of reflexive and leaflet processes. At the interfaces, varying sizes of astrocyte-astrocyte contacts were identified. Inside an astrocyte domain, synapses contact the entire astrocyte, and synapse-astrocyte contacts increase from soma to terminal leaflets. In contrast to densely packed vesicles at synaptic boutons, vesicle-like structures were scant within astrocytes. Lastly, astrocytes contain dense mitochondrial networks with a mitochondrial volume ratio similar to that of neurites. Together, these ultrastructural details should expand our understanding of functional astrocyte-astrocyte and astrocyte-neuron interactions.


Asunto(s)
Astrocitos , Sinapsis , Astrocitos/metabolismo , Encéfalo , Humanos , Mitocondrias , Neuronas/fisiología , Sinapsis/metabolismo
5.
J Neurophysiol ; 126(4): 1403-1419, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34525325

RESUMEN

Predominant expression of leak-type K+ channels provides astrocytes a high membrane permeability to K+ ions and a hyperpolarized membrane potential that are crucial for astrocyte function in brain homeostasis. In functionally mature astrocytes, the expression of leak K+ channels creates a unique membrane K+ conductance that lacks voltage-dependent rectification. Accordingly, the conductance is named ohmic or passive K+ conductance. Several inwardly rectifying and two-pore domain K+ channels have been investigated for their contributions to passive conductance. Meanwhile, gap junctional coupling has been postulated to underlie the passive behavior of membrane conductance. It is now clear that the intrinsic properties of K+ channels and gap junctional coupling can each act alone or together to bring about a passive behavior of astrocyte conductance. Additionally, while the passive conductance can generally be viewed as a K+ conductance, the actual representation of this conductance is a combined expression of multiple known and unknown K+ channels, which has been further modified by the intricate morphology of individual astrocytes and syncytial gap junctional coupling. The expression of the inwardly rectifying K+ channels explains the inward-going component of passive conductance disobeying Goldman-Hodgkin-Katz constant field outward rectification. However, the K+ channels encoding the outward-going passive currents remain to be determined in the future. Here, we review our current understanding of ion channels and biophysical mechanisms engaged in the passive astrocyte K+ conductance, propose new studies to resolve this long-standing puzzle in astrocyte physiology, and discuss the functional implication(s) of passive behavior of K+ conductance on astrocyte physiology.


Asunto(s)
Astrocitos/fisiología , Fenómenos Biofísicos/fisiología , Uniones Comunicantes/fisiología , Potenciales de la Membrana/fisiología , Canales de Potasio/fisiología , Potasio/metabolismo , Animales , Humanos
6.
Brain Sci ; 10(4)2020 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-32252295

RESUMEN

Astrocyte syncytial isopotentiality is a physiological mechanism resulting from a strong electrical coupling among astrocytes. We have previously shown that syncytial isopotentiality exists as a system-wide feature that coordinates astrocytes into a system for high efficient regulation of brain homeostasis. Neuronal activity is known to regulate gap junction coupling through alteration of extracellular ions and neurotransmitters. However, the extent to which epileptic neuronal activity impairs the syncytial isopotentiality is unknown. Here, the neuronal epileptiform bursts were induced in acute hippocampal slices by removal of Mg2+ (Mg2+ free) from bath solution and inhibition of γ-aminobutyric acid A (GABAA) receptors by 100 µM picrotoxin (PTX). The change in syncytial coupling was monitored by using a K+ free-Na+-containing electrode solution ([Na+]p) in the electrophysiological recording where the substitution of intracellular K+ by Na+ ions dissipates the physiological membrane potential (VM) to ~0 mV in the recorded astrocyte. However, in a syncytial coupled astrocyte, the [Na+]p induced VM loss can be compensated by the coupled astrocytes to a quasi-physiological membrane potential of ~73 mV. After short-term exposure to this experimental epileptic condition, a significant closure of syncytial coupling was indicated by a shift of the quasi-physiological membrane potential to -60 mV, corresponding to a 90% reduction of syncytial coupling strength. Consequently, the closure of syncytial coupling significantly decreased the ability of the syncytium for spatial redistribution of K+ ions. Altogether, our results show that epileptiform neuronal discharges weaken the strength of syncytial coupling and that in turn impairs the capacity of a syncytium for spatial redistribution of K+ ions.

7.
Mol Neurobiol ; 57(3): 1332-1346, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31728930

RESUMEN

TREK-1, a two-pore-domain K+ channel, is highly expressed in the central nervous system. Although aberrant expression of TREK-1 is implicated in cognitive impairment, the cellular and functional mechanism underlying this channelopathy is poorly understood. Here we examined TREK-1 contribution to neuronal morphology, excitability, synaptic plasticity, and cognitive function in mice deficient in TREK-1 expression. TREK-1 immunostaining signal mainly appeared in hippocampal pyramidal neurons, but not in astrocytes. TREK-1 gene knockout (TREK-1 KO) increases dendritic sprouting and the number of immature spines in hippocampal CA1 pyramidal neurons. Functionally, TREK-1 KO increases neuronal excitability and enhances excitatory and inhibitory postsynaptic currents (EPSCs and IPSCs). The increased EPSCs appear to be attributed to an increased release probability of presynaptic glutamate and functional expression of postsynaptic AMPA receptors. TREK-1 KO decreased the paired-pulse ratio and severely occluded the long-term potentiation (LTP) in the CA1 region. These altered synaptic transmission and plasticity are associated with recognition memory deficit in TREK-1 KO mice. Although astrocytic expression of TREK-1 has been reported in previous studies, TREK-1 KO does not alter astrocyte membrane K+ conductance or the syncytial network function in terms of syncytial isopotentiality. Altogether, TREK-1 KO profoundly affects the cellular structure and function of hippocampal pyramidal neurons. Thus, the impaired cognitive function in diseases associated with aberrant expression of TREK-1 should be attributed to the failure of this K+ channel in regulating neuronal morphology, excitability, synaptic transmission, and plasticity.


Asunto(s)
Cognición/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Plasticidad Neuronal/genética , Neuronas/fisiología , Canales de Potasio de Dominio Poro en Tándem/genética , Animales , Astrocitos/metabolismo , Potenciales Postsinápticos Excitadores/genética , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Ratones Noqueados , Plasticidad Neuronal/fisiología , Células Piramidales/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
8.
Glia ; 66(12): 2756-2769, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30277621

RESUMEN

Syncytial isopotentiality, resulting from a strong electrical coupling, emerges as a physiological mechanism that coordinates individual astrocytes to function as a highly efficient system in brain homeostasis. However, whether syncytial isopotentiality occurs selectively to certain brain regions or is universal to astrocytic networks remains unknown. Here, we have explored the correlation of syncytial isopotentiality with different astrocyte subtypes in various brain regions. Using a nonphysiological K+ -free/Na+ electrode solution to depolarize a recorded astrocyte in situ, the existence of syncytial isopotentiality can be revealed: the recorded astrocyte's membrane potential remains at a quasi-physiological level due to strong electrical coupling with neighboring astrocytes. Syncytial isopotentiality appears in Layer I of the motor, sensory, and visual cortical regions, where astrocytes are organized with comparable cell densities, interastrocytic distances, and the quantity of directly coupled neighbors. Second, though astrocytes vary in their cytoarchitecture in association with neuronal circuits from Layers I-VI, the established syncytial isopotentiality remains comparable among different layers in the visual cortex. Third, neurons and astrocytes are uniquely organized as barrels in Layer IV somatosensory cortex; interestingly, astrocytes both inside and outside of the barrels do electrically communicate with each other and also share syncytial isopotentiality. Fourth, syncytial isopotentiality appears in radial-shaped Bergmann glia and velate astrocytes in the cerebellar cortex. Fifth, although fibrous astrocytes in white matter exhibit a distinct morphology, their network syncytial isopotentiality is comparable with protoplasmic astrocytes. Altogether, syncytial isopotentiality appears as a system-wide electrical feature of astrocytic networks in the brain.


Asunto(s)
Astrocitos/fisiología , Encéfalo/citología , Uniones Comunicantes/fisiología , Potenciales de la Membrana/fisiología , Red Nerviosa/fisiología , Familia de Aldehído Deshidrogenasa 1 , Animales , Animales Recién Nacidos , Células Cultivadas , Conexina 43/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Isoenzimas/genética , Isoenzimas/metabolismo , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp , Fosfopiruvato Hidratasa/metabolismo , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Sodio/metabolismo , Sustancia Blanca/citología
9.
Exp Neurol ; 303: 1-11, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29407729

RESUMEN

Membrane potential (VM) depolarization occurs immediately following cerebral ischemia and is devastating for the astrocyte homeostasis and neuronal signaling. Previously, an excessive release of extracellular K+ and glutamate has been shown to underlie an ischemia-induced VM depolarization. Ischemic insults should impair membrane ion channels and disrupt the physiological ion gradients. However, their respective contribution to ischemia-induced neuronal and glial depolarization and loss of neuronal excitability are unanswered questions. A short-term oxygen-glucose deprivation (OGD) was used for the purpose of examining the acute effect of ischemic conditions on ion channel activity and physiological K+ gradient in neurons and glial cells. We show that a 30 min OGD treatment exerted no measurable damage to the function of membrane ion channels in neurons, astrocytes, and NG2 glia. As a result of the resilience of membrane ion channels, neuronal spikes last twice as long as our previously reported 15 min time window. In the electrophysiological analysis, a 30 min OGD-induced dissipation of transmembrane K+ gradient contributed differently in brain cell depolarization: severe in astrocytes and neurons, and undetectable in NG2 glia. The discrete cellular responses to OGD corresponded to a total loss of 69% of the intracellular K+ contents in hippocampal slices as measured by Inductively Coupled Plasma Mass Spectrometry (ICP-MS). A major brain cell depolarization mechanism identified here is important for our understanding of cerebral ischemia pathology. Additionally, further understanding of the resilient response of NG2 glia to ischemia-induced intracellular K+ loss and depolarization should facilitate the development of future stroke therapy.


Asunto(s)
Astrocitos/fisiología , Fenómenos Biofísicos/fisiología , Glucosa/metabolismo , Hipoxia/fisiopatología , Potenciales de la Membrana/fisiología , Neuronas/fisiología , Potasio/metabolismo , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Fenómenos Biofísicos/efectos de los fármacos , Conductividad Eléctrica , Femenino , Células Gigantes/fisiología , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/farmacología , Técnicas de Placa-Clamp , Proteoglicanos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
10.
Mol Brain ; 9: 34, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27004553

RESUMEN

BACKGROUND: Neonatal astrocytes are diverse in origin, and undergo dramatic change in gene expression, morphological differentiation and  syncytial networking throughout development. Neonatal astrocytes also play multifaceted roles in neuronal circuitry establishment. However, the extent to which neonatal astrocytes differ from their counterparts in the adult brain remains unknown. RESULTS: Based on ALDH1L1-eGFP expression or sulforhodamine 101 staining, neonatal astrocytes at postnatal day 1-3 can be reliably identified in hippocampal stratum radiatum. They exhibit a more negative resting membrane potential (V M), -85 mV, than mature astrocytes, -80 mV and a variably rectifying whole-cell current profile due to complex expression of voltage-gated outward transient K(+) (IKa), delayed rectifying K(+) (IKd) and inward K(+) (IKin) conductances. Differing from NG2 glia, depolarization-induced inward Na(+) currents (INa) could not be detected in neonatal astrocytes. A quasi-physiological V M of -69 mV was retained when inwardly rectifying Kir4.1 was inhibited by 100 µM Ba(2+) in both wild type and TWIK-1/TREK-1 double gene knockout astrocytes, indicating expression of additional leak K(+) channels yet unknown. In dual patch recording, electrical coupling was detected in 74 % (14/19 pairs) of neonatal astrocytes with largely variable coupling coefficients. The increasing gap junction coupling progressively masked the rectifying K(+) conductances to account for an increasing number of linear voltage-to-current relationship passive astrocytes (PAs). Gap junction inhibition, by 100 µM meclofenamic acid, substantially reduced membrane conductance and converted all the neonatal PAs to variably rectifying astrocytes. The low density expression of leak K(+) conductance in neonatal astrocytes corresponded  to a ~50 % less K(+) uptake capacity compared to adult astrocytes. CONCLUSIONS: Neonatal astrocytes predominantly express a variety of rectifying K(+) conductances, form discrete cell-to-cell gap junction coupling and are deficient in K(+) homeostatic capacity.


Asunto(s)
Astrocitos/metabolismo , Fenómenos Electrofisiológicos , Hipocampo/metabolismo , Animales , Bario/metabolismo , Uniones Comunicantes/metabolismo , Activación del Canal Iónico , Cinética , Ratones Endogámicos C57BL , Fenotipo , Canales de Potasio de Rectificación Interna/metabolismo
11.
Front Cell Neurosci ; 10: 13, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26869883

RESUMEN

We have recently shown that a linear current-to-voltage (I-V) relationship of membrane conductance (passive conductance) reflects the intrinsic property of K(+) channels in mature astrocytes. While passive conductance is known to underpin a highly negative and stable membrane potential (V M) essential for the basic homeostatic function of astrocytes, a complete repertoire of the involved K(+) channels remains elusive. TREK-1 two-pore domain K(+) channel (K2P) is highly expressed in astrocytes, and covalent association of TREK-1 with TWIK-1, another highly expressed astrocytic K2P, has been reported as a mechanism underlying the trafficking of heterodimer TWIK-1/TREK-1 channel to the membrane and contributing to astrocyte passive conductance. To decipher the individual contribution of TREK-1 and address whether the appearance of passive conductance is conditional to the co-expression of TWIK-1/TREK-1 in astrocytes, TREK-1 single and TWIK-1/TREK-1 double gene knockout mice were used in the present study. The relative quantity of mRNA encoding other astrocyte K(+) channels, such as Kir4.1, Kir5.1, and TREK-2, was not altered in these gene knockout mice. Whole-cell recording from hippocampal astrocytes in situ revealed no detectable changes in astrocyte passive conductance, V M, or membrane input resistance (R in) in either kind of gene knockout mouse. Additionally, TREK-1 proteins were mainly located in the intracellular compartments of the hippocampus. Altogether, genetic deletion of TREK-1 alone or together with TWIK-1 produced no obvious alteration in the basic electrophysiological properties of hippocampal astrocytes. Thus, future research focusing on other K(+) channels may shed light on this long-standing and important question in astrocyte physiology.

12.
J Thorac Dis ; 8(1): E8-E19, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26904258

RESUMEN

Air pollution is now becoming an independent risk factor for cardiovascular morbidity and mortality. Numerous epidemiological, biomedical and clinical studies indicate that ambient particulate matter (PM) in air pollution is strongly associated with increased cardiovascular disease such as myocardial infarction (MI), cardiac arrhythmias, ischemic stroke, vascular dysfunction, hypertension and atherosclerosis. The molecular mechanisms for PM-caused cardiovascular disease include directly toxicity to cardiovascular system or indirectly injury by inducing systemic inflammation and oxidative stress in peripheral circulation. Here, we review the linking between PM exposure and the occurrence of cardiovascular disease and discussed the possible underlying mechanisms for the observed PM induced increases in cardiovascular morbidity and mortality.

13.
Mol Neurobiol ; 53(9): 6169-6182, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26553349

RESUMEN

TWIK-1 two-pore domain K+ channels are highly expressed in mature hippocampal astrocytes. While the TWIK-1 activity is readily detectable on astrocyte membrane, the majority of channels are retained in the intracellular compartments, which raises an intriguing question of whether the membrane TWIK-1 channels could be dynamically regulated for functions yet unknown. Here, the regulation of TWIK-1 membrane expression by Gi/Go-coupled metabotropic glutamate receptor 3 (mGluR3) and its functional impact on ammonium uptake has been studied. Activation of mGluR3 induced a marked translocation of TWIK-1 channels from the cytoplasm to the membrane surface. Consistent with our early observation that membrane TWIK-1 behaves as nonselective monovalent cation channel, mGluR3-mediated TWIK-1 membrane expression was associated with a depolarizing membrane potential (V M). As TWIK-1 exhibits a discernibly high permeability to ammonium (NH4+), a critical substrate in glutamate-glutamine cycle for neurotransmitter replenishment, regulation of NH4+ uptake capacity by TWIK-1 membrane expression was determined by response of astrocyte V M to bath application of 5 mM NH4Cl. Stimulation of mGluR3 potentiated NH4+-induced V M depolarization by ∼30 % in wild type, but not in TWIK-1 knockout astrocytes. Furthermore, activation of mGluR3 mediated a coordinated translocation of TWIK-1 channels with recycling endosomes toward astrocyte membrane and the mGluR3-mediated potentiation of NH4+ uptake required a functional Rab-mediated trafficking pathway. Altogether, we demonstrate that the activation of mGluR3 up-regulates the membrane expression of TWIK-1 that in turn enhances NH4+ uptake in astrocytes, a mechanism potentially important for functional coupling of astrocyte glutamate-glutamine cycle with the replenishment of neurotransmitters in neurons.


Asunto(s)
Compuestos de Amonio/metabolismo , Astrocitos/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Hipocampo/citología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Endocitosis , Endosomas/metabolismo , Exocitosis , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Glutamato Metabotrópico/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Proteínas de Unión al GTP rab/metabolismo
14.
Glia ; 64(2): 214-26, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26435164

RESUMEN

Astrocytes are extensively coupled through gap junctions into a syncytium. However, the basic role of this major brain network remains largely unknown. Using electrophysiological and computational modeling methods, we demonstrate that the membrane potential (VM) of an individual astrocyte in a hippocampal syncytium, but not in a single, freshly isolated cell preparation, can be well-maintained at quasi-physiological levels when recorded with reduced or K(+) free pipette solutions that alter the K(+) equilibrium potential to non-physiological voltages. We show that an astrocyte's associated syncytium provides powerful electrical coupling, together with ionic coupling at a lesser extent, that equalizes the astrocyte's VM to levels comparable to its neighbors. Functionally, this minimizes VM depolarization attributable to elevated levels of local extracellular K(+) and thereby maintains a sustained driving force for highly efficient K(+) uptake. Thus, gap junction coupling functions to achieve isopotentiality in astrocytic networks, whereby a constant extracellular environment can be powerfully maintained for crucial functions of neural circuits.


Asunto(s)
Astrocitos/fisiología , Uniones Comunicantes/fisiología , Potenciales de la Membrana/fisiología , Animales , Cationes Monovalentes/metabolismo , Células Cultivadas , Espacio Extracelular/metabolismo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/fisiología , Potasio/metabolismo , Técnicas de Cultivo de Tejidos
15.
J Neurophysiol ; 113(10): 3744-50, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810481

RESUMEN

Mature astrocytes exhibit a linear current-to-voltage K(+) membrane conductance (passive conductance) and an extremely low membrane resistance (Rm) in situ. The combination of these electrophysiological characteristics establishes a highly negative and stable membrane potential that is essential for basic functions, such as K(+) spatial buffering and neurotransmitter uptake. However, astrocytes are coupled extensively in situ. It remains to be determined whether the observed passive behavior and low Rm are attributable to the intrinsic properties of membrane ion channels or to gap junction coupling in functionally mature astrocytes. In the present study, freshly dissociated hippocampal tissues were used as a new model to examine this basic question in young adult animals. The morphologically intact single astrocytes could be reliably dissociated from animals postnatal day 21 and older. At this animal age, dissociated single astrocytes exhibit passive conductance and resting membrane potential similar to those exhibited by astrocytes in situ. To precisely measure the Rm from single astrocytes, dual-patch single-astrocyte recording was performed. We show that dissociated single astrocytes exhibit a low Rm similarly to syncytial coupled astrocytes. Functionally, the symmetric expression of high-K(+) conductance enabled rapid change in the intracellular K(+) concentrations in response to changing K(+) drive force. Altogether, we demonstrate that freshly dissociated tissue preparation is a highly useful model for study of the functional expression and regulation of ion channels, receptors, and transporters in astrocytes and that passive behavior and low Rm are the intrinsic properties of mature astrocytes.


Asunto(s)
Astrocitos/fisiología , Uniones Comunicantes/fisiología , Hipocampo/citología , Potenciales de la Membrana/fisiología , Animales , Biofisica , Estimulación Eléctrica , Técnicas In Vitro , Ratones , Microscopía Confocal , Técnicas de Placa-Clamp , Potasio/metabolismo , Rodaminas/metabolismo
16.
J Thorac Dis ; 6(5): 477-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24822106

RESUMEN

OBJECTIVE: To examine the association between hemoglobin (Hb) levels and cardiovascular risk factors in a large community-dwelling cohort. METHODS: A total of 4,186 women and 4,851 men were enrolled in the study. Data on personal history, physical examination and biochemical parameters were collected. Subjects were categorized by gender and divided into different group according to the level of Hb or blood pressure, and the association between Hb levels and cardiovascular risk factors was examined using Pearson's correlation analysis. RESULTS: In both men and women even with normal Hb level, tertiles of Hb levels were positively associated with body mass index (BMI), total-cholesterol (TC), triglyceride (TG), uric acid (UA), diastolic blood pressures (DBP) and fasting plasma glucose (FPG) (all P=0.000 in men and women). Furthermore, significantly increased incidence of hyperuricemia (P=0.000 both in men and women) and obesity (P=0.000 both in men and women) were observed with the gradually increased Hb level. In addition, Pearson's correlation analysis revealed obvious correlation between Hb level and various cardiovascular risk factors including blood pressure and UA. Binary logistic regression analysis further demonstrated that the level of Hb was an important risk factor for elevated blood pressure (OR =1.216; 95% CI: 1.138-1.293, P=0.000 in men; OR =1.287; 95% CI: 1.229-1.363, P=0.000 in women). CONCLUSIONS: Increasing Hb levels, even in subjects with normal level were associated with increasing prevalence of cardiovascular risk factors, suggesting that a slightly low Hb level might be beneficial to Chinese community-dwelling individuals.

17.
Ann Neurol ; 70(1): 121-32, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21786302

RESUMEN

OBJECTIVE: Most patients with stroke caused by middle cerebral artery occlusion (MCAO) show cognitive deficit that is generally regarded as resulting from damage to the cerebral cortex rather than the hippocampus. Whether MCAO induces hippocampal damage and whether this contributes to the cognitive defects remains unclear. Here we investigate the hippocampal damage and its correlation to cognitive defects after exclusively unilateral MCAO and the underlying mechanism for that damage. METHODS: Patients were assessed for hippocampal damage by magnetic resonance imaging (MRI) and magnetic resonance angiography (MRA), and the Mini Mental-Status Evaluation (MMSE) and Rey Auditory Verbal Learning Test (RAVLT) were used to assess for cognitive defects. RESULTS: We provide the first evidence that patients with exclusively unilateral MCAO showed hippocampal damage characterized by an infarct-size-independent atrophy and alterations in neuronal and glial metabolites in the ipsilateral hippocampus, in parallel with cognitive impairment. Rodent MCAO also induced delayed shrinkage and pyramidal neuronal death in the ipsilateral hippocampus and an impairment of hippocampal-dependent spatial memory. Blocking Gap junctional communication (GJC) with carbenoxolone or downregulation of connexin43 (Cx43) significantly increased the survival of the pyramidal neurons in the ipsilateral hippocampus and improved behavioral scores. Furthermore, Cx43 heterozygous mice showed reduced shrinkage and metabolite abnormality in ipsilateral hippocampus after MCAO. INTERPRETATION: Astroglial GJC plays a significant role in MCAO-induced remote hippocampal damage and cognitive impairment. It might be possible to improve the cognition in patients with MCAO by manipulating interastrocytic communication via the gap junction channels.


Asunto(s)
Comunicación Celular , Uniones Comunicantes/patología , Hipocampo/patología , Infarto de la Arteria Cerebral Media/patología , Neuroglía/patología , Adulto , Animales , Comunicación Celular/fisiología , Femenino , Uniones Comunicantes/metabolismo , Hipocampo/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Neuroglía/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...